Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.834
Filtrer
1.
Front Immunol ; 15: 1402123, 2024.
Article de Anglais | MEDLINE | ID: mdl-39086482

RÉSUMÉ

Background: Control of buffalo flies (Haematobia irritans exigua, BFs) relies mainly on chemical methods; however, resistance to insecticides is widespread in BF populations. Breeding for resistance to BFs represents a possible alternative, but direct phenotyping of animals is laborious and often inaccurate. The availability of reliable diagnostic biomarker(s) to identify low BF carrier cattle would facilitate rapid and accurate selection for genetic improvement. However, limited information is available regarding differences amongst cattle in host responses to BF infestation. Methods: This study investigated the variation in Brangus cattle serum proteomic profiles before (naïve) and after peak BF exposure, in low (LF) and high BF burden (HF) cattle. Cattle were phenotyped for susceptibility based on BF counts on multiple dates using visual and photographic techniques. The relative abundance of serum proteins in cattle before and after exposure to BFs was analysed using sequential window acquisition of all theoretical fragment ion mass spectrometry (SWATH-MS). Results: Exposure to BFs elicited similar responses in HF and LF cattle, with 79 and 70 proteins, respectively, showing significantly different abundances post exposure as compared to their relevant naïve groups. The comparison of serum samples from naïve HF and LF cattle identified 44 significantly differentially abundant (DA) proteins, while 37 significantly DA proteins were identified from the comparison between HF and LF cattle post-exposure to BFs. Proteins with higher abundance in naïve LF cattle were enriched in blood coagulation mechanisms that were sustained after exposure to BFs. Strong immune response mechanisms were also identified in naïve LF cattle, whereas these responses developed in HF cattle only after exposure to BF. High BF cattle also showed active anticoagulation mechanisms in response to BF exposure, including downregulation of coagulation factor IX and upregulation of antithrombin-III, which might facilitate BF feeding. Conclusion: Underlying differences in the abundance of proteins related to blood coagulation and immune response pathways could potentially provide indirect indicators of susceptibility to BF infestation and biomarkers for selecting more BF-resistant cattle.


Sujet(s)
Protéomique , Animaux , Bovins , Protéomique/méthodes , Prédisposition aux maladies , Maladies des bovins/immunologie , Maladies des bovins/sang , Maladies des bovins/parasitologie , Marqueurs biologiques/sang , Myiases/médecine vétérinaire , Myiases/immunologie , Interactions hôte-parasite/immunologie , Protéines du sang/métabolisme , Protéines du sang/analyse , Protéome
2.
Immunity ; 57(8): 1726-1728, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39142272

RÉSUMÉ

Understanding determinants of immune response variation is central to developing treatment options. Even et al. show that naive CD4+ T cell transcriptional heterogeneity is altered by helminth infection leading to impaired immune responses independent of commensals.


Sujet(s)
Lymphocytes T CD4+ , Helminthes , Animaux , Humains , Helminthes/immunologie , Lymphocytes T CD4+/immunologie , Helminthiase/immunologie , Helminthiase/parasitologie , Interactions hôte-parasite/immunologie , Souris
3.
Front Immunol ; 15: 1392569, 2024.
Article de Anglais | MEDLINE | ID: mdl-38983863

RÉSUMÉ

Introduction: Parasite-mediated selection is considered one of the potential mechanisms contributing to the coexistence of asexual-sexual complexes. Gibel carp (Carassius gibelio), an invasive fish species in Europe, often forms populations composed of gynogenetic and sexual specimens. Methods: The experimental infection was induced in gynogenetic and sexual gibel carp using eye-fluke Diplostomum pseudospathaceum (Trematoda), and the transcriptome profile of the spleen as a major immune organ in fish was analyzed to reveal the differentially expressed immunity-associated genes related to D. pseudospathaceum infection differing between gynogenetic and sexual gibel carp. Results: High parasite infection was found in gynogenetic fish when compared to genetically diverse sexuals. Although metacercariae of D. pseudospathaceum are situated in an immune-privileged organ, our results show that eye trematodes may induce a host immune response. We found differential gene expression induced by eye-fluke infection, with various impacts on gynogenetic and sexual hosts, documenting for the majority of DEGs upregulation in sexuals, and downregulation in asexuals. Differences in gene regulation between gynogenetic and sexual gibel carp were evidenced in many immunity-associated genes. GO analyses revealed the importance of genes assigned to the GO terms: immune function, the Notch signaling pathway, MAP kinase tyrosine/threonine/phosphatase activity, and chemokine receptor activity. KEGG analyses revealed the importance of the genes involved in 12 immunity-associated pathways - specifically, FoxO signaling, adipocytokine signaling, TGF-beta signaling, apoptosis, Notch signaling, C-type lectin receptor signaling, efferocytosis, intestinal immune network for IgA production, insulin signaling, virion - human immunodeficiency virus, Toll-like receptor signaling, and phosphatidylinositol signaling system. Discussion: Our study indicates the limited potential of asexual fish to cope with higher parasite infection (likely a loss of capacity to induce an effective immune response) and highlights the important role of molecular mechanisms associated with immunity for the coexistence of gynogenetic and sexual gibel carp, potentially contributing to its invasiveness.


Sujet(s)
Maladies des poissons , Espèce introduite , Trematoda , Infections à trématodes , Animaux , Trematoda/physiologie , Maladies des poissons/immunologie , Maladies des poissons/parasitologie , Infections à trématodes/médecine vétérinaire , Infections à trématodes/immunologie , Infections à trématodes/parasitologie , Transcriptome , Interactions hôte-parasite/immunologie , Régulation de l'expression des gènes , Carpes (poisson)/parasitologie , Carpes (poisson)/immunologie , Carpes (poisson)/génétique , Femelle , Analyse de profil d'expression de gènes , Reproduction/immunologie
4.
Front Immunol ; 15: 1428232, 2024.
Article de Anglais | MEDLINE | ID: mdl-39040112

RÉSUMÉ

In the decades since the discovery, Type I interferon (IFN-I) has been intensively studied for their antiviral activity. However, increasing evidences suggest that it may also play an important role in the infection of Toxoplasma gondii, a model organism for intracellular parasites. Recent studies demonstrated that the induction of IFN-I by the parasite depends on cell type, strain genotype, and mouse strain. IFN-I can inhibit the proliferation of T. gondii, but few studies showed that it is beneficial to the growth of the parasite. Meanwhile, T. gondii also can secrete proteins that impact the pathway of IFN-I production and downstream induced interferon-stimulated genes (ISGs) regulation, thereby escaping immune destruction by the host. This article reviews the major findings and progress in the production, function, and regulation of IFN-I during T. gondii infection, to thoroughly understand the innate immune mechanism of T. gondii infection, which provides a new target for subsequent intervention and treatment.


Sujet(s)
Interféron de type I , Toxoplasma , Toxoplasmose , Toxoplasma/immunologie , Animaux , Interféron de type I/immunologie , Interféron de type I/métabolisme , Humains , Toxoplasmose/immunologie , Toxoplasmose/parasitologie , Interactions hôte-parasite/immunologie , Immunité innée , Transduction du signal , Régulation de l'expression des gènes , Souris
6.
Trends Parasitol ; 40(7): 549-561, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38853079

RÉSUMÉ

Helminths can adapt to environmental conditions in the host, utilising anaerobic processes like fermentation and malate dismutation to produce energy from carbohydrate. Although targeting carbohydrate metabolism is an established therapeutic strategy to combat helminth infection, questions remain over the metabolic pathways they employ as adults to survive and evade host immunity. Helminths also use amino acid, polyunsaturated fatty acid (PUFA), and cholesterol metabolism, a possible strategy favouring the production of immunomodulatory compounds that may influence survival in the host. Here, we discuss the significance of these differing metabolic pathways and whether targeting of helminth metabolic pathways may allow for the development of novel anthelmintics.


Sujet(s)
Helminthiase , Helminthes , Interactions hôte-parasite , Animaux , Helminthes/immunologie , Helminthes/physiologie , Interactions hôte-parasite/immunologie , Interactions hôte-parasite/physiologie , Helminthiase/immunologie , Helminthiase/parasitologie , Humains , Anthelminthiques/usage thérapeutique , Anthelminthiques/pharmacologie
7.
Nat Commun ; 15(1): 4913, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851821

RÉSUMÉ

Host immune responses are tightly controlled by various immune factors during infection, and protozoan parasites also manipulate the immune system to evade surveillance, leading to an evolutionary arms race in host‒pathogen interactions; however, the underlying mechanisms are not fully understood. We observed that the level of superoxide dismutase 3 (SOD3) was significantly elevated in both Plasmodium falciparum malaria patients and mice infected with four parasite species. SOD3-deficient mice had a substantially longer survival time and lower parasitemia than control mice after infection, whereas SOD3-overexpressing mice were much more vulnerable to parasite infection. We revealed that SOD3, secreted from activated neutrophils, bound to T cells, suppressed the interleukin-2 expression and concomitant interferon-gamma responses crucial for parasite clearance. Overall, our findings expose active fronts in the arms race between the parasites and host immune system and provide insights into the roles of SOD3 in shaping host innate immune responses to parasite infection.


Sujet(s)
Paludisme à Plasmodium falciparum , Souris de lignée C57BL , Souris knockout , Granulocytes neutrophiles , Superoxide dismutase , Animaux , Superoxide dismutase/métabolisme , Superoxide dismutase/génétique , Humains , Souris , Granulocytes neutrophiles/immunologie , Paludisme à Plasmodium falciparum/immunologie , Paludisme à Plasmodium falciparum/parasitologie , Immunité cellulaire , Lymphocytes T/immunologie , Plasmodium falciparum/immunologie , Femelle , Interactions hôte-parasite/immunologie , Interactions hôte-parasite/génétique , Interféron gamma/métabolisme , Interféron gamma/immunologie , Mâle , Immunité innée , Interleukine-2/métabolisme , Interleukine-2/immunologie , Interleukine-2/génétique , Parasitémie/immunologie
8.
Microbiol Spectr ; 12(8): e0047224, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-38888326

RÉSUMÉ

Diarrheal diseases with infectious etiology remain a major cause of death globally, particularly in low-income countries. Entamoeba histolytica is a pathogenic protozoan parasite that is the causative agent of amebiasis. Amebiasis has a wide presentation in clinical severity with many factors, including the bacterial microbiota, contributing to this variation. The innate immune response also plays a critical role in regulating the severity of E. histolytica infection, with neutrophils reported to have a protective role. Despite this, the precise mechanism of how neutrophils mediate amebic killing is poorly understood. Thus, modern platforms that allow for inquiry of granulocyte-ameba interactions will increase our understanding of this disease. Herein, we describe an assay for neutrophil killing of E. histolytica by utilizing high-dimensional spectral flow cytometry. Neutrophils were isolated from wild-type 5-week-old C57BL/6 mice and co-cultured with E. histolytica at various multiplicity of infections (MOIs). After co-culture, neutrophils and E. histolytica were stained for spectral flow cytometry. Cell populations were identified using surface markers and fluorescence minus one (FMO) controls. We have previously shown that animals colonized with a component of the human microbiota, Clostridium scindens, were protected from E. histolytica. This protection was associated with elevated neutrophil count. Here, we explored amebic killing capacity and observed that neutrophils from animals with C. scindens possessed heightened amebic killing compared with controls. Thus, this study establishes a novel platform that can provide an in-depth analysis of granulocyte-parasite interactions in various contexts, including during alteration of the intestinal microbiota.IMPORTANCEThe tools for studying host immune cell-E. histolytica interactions are limited. Factors, such as parasite heterogeneity, infectivity, and difficulties with culture systems and animal models, make interrogation of these interactions challenging. Thus, Entamoeba researchers can benefit from next-generation models that allow for the analysis of both host and parasite cells. Here, we demonstrate the use of a novel platform that allows for the determination of parasite-host cell interactions and customizable high-dimensional phenotyping of both populations. Indeed, spectral flow cytometry can approach >40 markers on a single panel and can be paired with custom-developed parasite antibodies that can be conjugated to fluorochromes via commercially available kits. This platform affords researchers the capability to test highly precise hypotheses regarding host-parasite interactions.


Sujet(s)
Entamoeba histolytica , Cytométrie en flux , Souris de lignée C57BL , Granulocytes neutrophiles , Animaux , Granulocytes neutrophiles/immunologie , Souris , Entamoeba histolytica/immunologie , Interactions hôte-parasite/immunologie , Humains , Infection à Entamoeba/immunologie , Infection à Entamoeba/parasitologie
9.
Adv Parasitol ; 124: 1-55, 2024.
Article de Anglais | MEDLINE | ID: mdl-38754926

RÉSUMÉ

Intestinal trematodes constitute a major group of helminths that parasitize humans and animals with relevant morbidity and mortality. Despite the importance of the intestinal trematodes in medical and veterinary sciences, immunology and pathology of these helminth infections have been neglected for years. Apart from the work focused on the members of the family Echnistomatidae, there are only very isolated and sporadic studies on the representatives of other families of digeneans, which makes a compilation of all these studies necessary. In the present review, the most salient literature on the immunology and pathology of intestinal trematodes in their definitive hosts in examined. Emphasis will be placed on members of the echinostomatidae family, since it is the group in which the most work has been carried out. However, we also review the information on selected species of the families Brachylaimidae, Diplostomidae, Gymnophallidae, and Heterophyidae. For most of these families, coverage is considered under the following headings: (i) Background; (ii) Pathology of the infection; (iii) Immunology of the infection; and (iv) Human infections.


Sujet(s)
Parasitoses intestinales , Trematoda , Infections à trématodes , Animaux , Humains , Trematoda/physiologie , Trematoda/immunologie , Infections à trématodes/parasitologie , Infections à trématodes/immunologie , Infections à trématodes/médecine vétérinaire , Parasitoses intestinales/immunologie , Parasitoses intestinales/parasitologie , Intestins/parasitologie , Intestins/anatomopathologie , Intestins/immunologie , Interactions hôte-parasite/immunologie
10.
Int Immunopharmacol ; 134: 112100, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38728877

RÉSUMÉ

The parasite Leishmania resides as amastigotes within the macrophage parasitophorous vacuoles inflicting the disease Leishmaniasis. Leishmania selectively modulates mitogen-activated protein kinase (MAPK) phosphorylation subverting CD40-triggered anti-leishmanial functions of macrophages. The mechanism of any pathogen-derived molecule induced host MAPK modulation remains poorly understood. Herein, we show that of the fifteen MAPKs, LmjMAPK4 expression is higher in virulent L. major. LmjMAPK4- detected in parasitophorous vacuoles and cytoplasm- binds MEK-1/2, but not MKK-3/6. Lentivirally-overexpressed LmjMAPK4 augments CD40-activated MEK-1/2-ERK-1/2-MKP-1, but inhibits MKK3/6-p38MAPK-MKP-3, phosphorylation. A rationally-identified LmjMAPK4 inhibitor reinstates CD40-activated host-protective anti-leishmanial functions in L. major-infected susceptible BALB/c mice. These results identify LmjMAPK4 as a MAPK modulator at the host-pathogen interface and establish a pathogen-intercepted host receptor signaling as a scientific rationale for identifying drug targets.


Sujet(s)
Antigènes CD40 , Leishmania major , Leishmaniose cutanée , Macrophages , Souris de lignée BALB C , Transduction du signal , Animaux , Leishmania major/immunologie , Leishmania major/physiologie , Antigènes CD40/métabolisme , Souris , Leishmaniose cutanée/immunologie , Leishmaniose cutanée/parasitologie , Macrophages/immunologie , Macrophages/parasitologie , Humains , Femelle , Phosphorylation , Interactions hôte-parasite/immunologie , Système de signalisation des MAP kinases/immunologie
11.
PeerJ ; 12: e17348, 2024.
Article de Anglais | MEDLINE | ID: mdl-38770098

RÉSUMÉ

Lake Baikal is one of the largest and oldest freshwater reservoirs on the planet with a huge endemic diversity of amphipods (Amphipoda, Crustacea). These crustaceans have various symbiotic relationships, including the rarely described phenomenon of leech parasitism on amphipods. It is known that leeches feeding on hemolymph of crustacean hosts can influence their physiology, especially under stressful conditions. Here we show that leeches Baicalobdella torquata (Grube, 1871) found on gills of Eulimnogammarus verrucosus (Gerstfeldt, 1858), one of the most abundant amphipods in the Baikal littoral zone, indeed feed on the hemolymph of their host. However, the leech infection had no effect on immune parameters such as hemocyte concentration or phenoloxidase activity and also did not affect glycogen content. The intensity of hemocyte reaction to foreign bodies in a primary culture was identical between leech-free and leech-infected animals. Artificial infection with leeches also had only a subtle effect on the course of a model microbial infection in terms of hemocyte concentration and composition. Despite we cannot fully exclude deleterious effects of the parasites, our study indicates a low influence of a few leeches on E. verrucosus and shows that leech-infected amphipods can be used at least for some types of ecophysiological experiments.


Sujet(s)
Amphipoda , Hémocytes , Hémolymphe , Lacs , Sangsues , Animaux , Amphipoda/immunologie , Amphipoda/parasitologie , Hémolymphe/immunologie , Hémolymphe/parasitologie , Sangsues/immunologie , Lacs/parasitologie , Hémocytes/immunologie , Immunité cellulaire , Sibérie , Interactions hôte-parasite/immunologie
12.
Parasit Vectors ; 17(1): 203, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38711063

RÉSUMÉ

BACKGROUND: The role of pathogen genotype in determining disease severity and immunopathology has been studied intensively in microbial pathogens including bacteria, fungi, protozoa and viruses but is poorly understood in parasitic helminths. The medically important blood fluke Schistosoma mansoni is an excellent model system to study the impact of helminth genetic variation on immunopathology. Our laboratory has demonstrated that laboratory schistosome populations differ in sporocyst growth and cercarial production in the intermediate snail host and worm establishment and fecundity in the vertebrate host. Here, we (i) investigate the hypothesis that schistosome genotype plays a significant role in immunopathology and related parasite life history traits in the vertebrate mouse host and (ii) quantify the relative impact of parasite and host genetics on infection outcomes. METHODS: We infected BALB/c and C57BL/6 mice with four different laboratory schistosome populations from Africa and the Americas. We quantified disease progression in the vertebrate host by measuring body weight and complete blood count (CBC) with differential over a 12-week infection period. On sacrifice, we assessed parasitological (egg and worm counts, fecundity), immunopathological (organ measurements and histopathology) and immunological (CBC with differential and cytokine profiles) characteristics to determine the impact of parasite and host genetics. RESULTS: We found significant variation between parasite populations in worm numbers, fecundity, liver and intestine egg counts, liver and spleen weight, and fibrotic area but not in granuloma size. Variation in organ weight was explained by egg burden and intrinsic parasite factors independent of egg burden. We found significant variation between infected mouse lines in cytokine levels (IFN-γ, TNF-α), eosinophils, lymphocytes and monocyte counts. CONCLUSIONS: This study showed that both parasite and host genotype impact the outcome of infection. While host genotype explains most of the variation in immunological traits, parasite genotype explains most of the variation in parasitological traits, and both host and parasite genotypes impact immunopathology outcomes.


Sujet(s)
Génotype , Souris de lignée BALB C , Souris de lignée C57BL , Schistosoma mansoni , Schistosomiase à Schistosoma mansoni , Animaux , Schistosoma mansoni/immunologie , Schistosoma mansoni/génétique , Souris , Schistosomiase à Schistosoma mansoni/immunologie , Schistosomiase à Schistosoma mansoni/parasitologie , Schistosomiase à Schistosoma mansoni/anatomopathologie , Femelle , Interactions hôte-parasite/immunologie , Interactions hôte-parasite/génétique , Cytokines/génétique , Cytokines/sang , Cytokines/immunologie
13.
Front Cell Infect Microbiol ; 14: 1369615, 2024.
Article de Anglais | MEDLINE | ID: mdl-38803570

RÉSUMÉ

Introduction: Little is known about the proteomic changes at the portals of entry in rainbow trout after infection with the myxozoan parasites, Myxobolus cerebralis, and Tetracapsuloides bryosalmonae. Whirling disease (WD) is a severe disease of salmonids, caused by the myxosporean M. cerebralis, while, proliferative kidney disease (PKD) is caused by T. bryosalmonae, which instead belongs to the class Malacosporea. Climate change is providing more suitable conditions for myxozoan parasites lifecycle, posing a high risk to salmonid aquaculture and contributing to the decline of wild trout populations in North America and Europe. Therefore, the aim of this study was to provide the first proteomic profiles of the host in the search for evasion strategies during single and coinfection with M. cerebralis and T. bryosalmonae. Methods: One group of fish was initially infected with M. cerebralis and another group with T. bryosalmonae. After 30 days, half of the fish in each group were co-infected with the other parasite. Using a quantitative proteomic approach, we investigated proteomic changes in the caudal fins and gills of rainbow trout before and after co-infection. Results: In the caudal fins, 16 proteins were differentially regulated post exposure to M. cerebralis, whereas 27 proteins were differentially modulated in the gills of the infected rainbow trout post exposure to T. bryosalmonae. After co-infection, 4 proteins involved in parasite recognition and the regulation of host immune responses were differentially modulated between the groups in the caudal fin. In the gills, 11 proteins involved in parasite recognition and host immunity, including 4 myxozoan proteins predicted to be virulence factors, were differentially modulated. Discussion: The results of this study increase our knowledge on rainbow trout co-infections by myxozoan parasites and rainbow trout immune responses against myxozoans at the portals of entry, supporting a better understanding of these host-parasite interactions.


Sujet(s)
Co-infection , Maladies des poissons , Myxobolus , Myxozoa , Oncorhynchus mykiss , Parasitoses animales , Protéomique , Animaux , Oncorhynchus mykiss/parasitologie , Oncorhynchus mykiss/immunologie , Maladies des poissons/parasitologie , Maladies des poissons/immunologie , Parasitoses animales/immunologie , Parasitoses animales/parasitologie , Co-infection/parasitologie , Co-infection/médecine vétérinaire , Co-infection/immunologie , Interactions hôte-parasite/immunologie , Protéome , Branchies/parasitologie , Branchies/immunologie , Branchies/métabolisme
14.
Front Immunol ; 15: 1370255, 2024.
Article de Anglais | MEDLINE | ID: mdl-38803499

RÉSUMÉ

Theileria equi (T. equi) is an apicomplexan parasite that causes severe hemolytic anemia in equids. Presently, there is inadequate knowledge of the immune responses induced by T. equi in equid hosts impeding understanding of the host parasite relationship and development of potent vaccines for control of T. equi infections. The objective of this study was to evaluate the host-parasite dynamics between T. equi merozoites and infected horses by assessing cytokine expression during primary and secondary parasite exposure, and to determine whether the pattern of expression correlated with clinical indicators of disease. Our findings showed that the expression of pro-inflammatory cytokines was very low and inconsistent during both primary and secondary infection. There was also no correlation between the symptoms observed during primary infection and expression of the cytokines. This suggests that the symptoms might have occurred primarily due to hemolysis and likely not the undesirable effects of pro-inflammatory responses. However, IL-10 and TGF-ß1 were highly expressed in both phases of infection, and their expression was linked to antibody production but not moderation of pro-inflammatory cytokine responses.


Sujet(s)
Maladies des chevaux , Interleukine-10 , Theileria , Theilériose , Facteur de croissance transformant bêta-1 , Animaux , Equus caballus , Theilériose/immunologie , Theilériose/parasitologie , Interleukine-10/métabolisme , Interleukine-10/immunologie , Theileria/immunologie , Facteur de croissance transformant bêta-1/métabolisme , Maladies des chevaux/immunologie , Maladies des chevaux/parasitologie , Mérozoïtes/immunologie , Anticorps antiprotozoaires/immunologie , Production d'anticorps/immunologie , Cytokines/métabolisme , Interactions hôte-parasite/immunologie
15.
Bioessays ; 46(7): e2400053, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38713161

RÉSUMÉ

Trypanosoma brucei is the causal agent of African Trypanosomiasis in humans and other animals. It maintains a long-term infection through an antigenic variation based population survival strategy. To proliferate in a mammal, T. brucei acquires iron and haem through the receptor mediated uptake of host transferrin and haptoglobin-hemoglobin respectively. The receptors are exposed to host antibodies but this does not lead to clearance of the infection. Here we discuss how the trypanosome avoids this fate in the context of recent findings on the structure and cell biology of the receptors.


Sujet(s)
Trypanosoma brucei brucei , Maladie du sommeil , Trypanosoma brucei brucei/immunologie , Trypanosoma brucei brucei/métabolisme , Humains , Animaux , Maladie du sommeil/immunologie , Maladie du sommeil/parasitologie , Haptoglobines/métabolisme , Récepteurs de surface cellulaire/métabolisme , Récepteurs de surface cellulaire/immunologie , Transferrine/métabolisme , Hémoglobines/métabolisme , Protéines de protozoaire/métabolisme , Protéines de protozoaire/immunologie , Interactions hôte-parasite/immunologie , Fer/métabolisme , Anticorps antiprotozoaires/immunologie
16.
BMC Biol ; 22(1): 89, 2024 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-38644510

RÉSUMÉ

BACKGROUND: Innate immune responses can be activated by pathogen-associated molecular patterns (PAMPs), danger signals released by damaged tissues, or the absence of self-molecules that inhibit immunity. As PAMPs are typically conserved across broad groups of pathogens but absent from the host, it is unclear whether they allow hosts to recognize parasites that are phylogenetically similar to themselves, such as parasitoid wasps infecting insects. RESULTS: Parasitoids must penetrate the cuticle of Drosophila larvae to inject their eggs. In line with previous results, we found that the danger signal of wounding triggers the differentiation of specialized immune cells called lamellocytes. However, using oil droplets to mimic infection by a parasitoid wasp egg, we found that this does not activate the melanization response. This aspect of the immune response also requires exposure to parasite molecules. The unidentified factor enhances the transcriptional response in hemocytes and induces a specific response in the fat body. CONCLUSIONS: We conclude that a combination of danger signals and the recognition of nonself molecules is required to activate Drosophila's immune response against parasitic insects.


Sujet(s)
Hémocytes , Interactions hôte-parasite , Immunité innée , Guêpes , Animaux , Guêpes/physiologie , Interactions hôte-parasite/immunologie , Hémocytes/immunologie , Drosophila melanogaster/parasitologie , Drosophila melanogaster/immunologie , Drosophila melanogaster/physiologie , Larve/immunologie , Larve/parasitologie , Drosophila/parasitologie , Drosophila/immunologie
17.
Fish Shellfish Immunol ; 149: 109580, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38663464

RÉSUMÉ

Wild organisms are regularly exposed to a wide range of parasites, requiring the management of an effective immune response while avoiding immunopathology. Currently, our knowledge of immunoparasitology primarily derives from controlled laboratory studies, neglecting the genetic and environmental diversity that contribute to immune phenotypes observed in wild populations. To gain insight into the immunologic variability in natural settings, we examined differences in immune gene expression of two Alaskan stickleback (Gasterosteus aculeatus) populations with varying susceptibility to infection by the cestode Schistocephalus solidus. Between these two populations, we found distinct immune gene expression patterns at the population level in response to infection with fish from the high-infection population displaying signs of parasite-driven immune manipulation. Further, we found significant differences in baseline immune gene profiles between the populations, with uninfected low-infection population fish showing signatures of inflammation compared to uninfected high-infection population fish. These results shed light on divergent responses of wild populations to the same parasite, providing valuable insights into host-parasite interactions in natural ecosystems.


Sujet(s)
Cestoda , Infections à cestodes , Maladies des poissons , Smegmamorpha , Animaux , Smegmamorpha/immunologie , Smegmamorpha/génétique , Smegmamorpha/parasitologie , Maladies des poissons/immunologie , Maladies des poissons/parasitologie , Infections à cestodes/médecine vétérinaire , Infections à cestodes/immunologie , Infections à cestodes/parasitologie , Cestoda/immunologie , Cestoda/physiologie , Interactions hôte-parasite/immunologie , Alaska , Immunité innée/génétique
18.
Trends Parasitol ; 40(5): 386-400, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38609741

RÉSUMÉ

Obesity is a worldwide pandemic and major risk factor for the development of metabolic syndrome (MetS) and type 2 diabetes (T2D). T2D requires lifelong medical support to limit complications and is defined by impaired glucose tolerance, insulin resistance (IR), and chronic low-level systemic inflammation initiating from adipose tissue. The current preventative strategies include a healthy diet, controlled physical activity, and medication targeting hyperglycemia, with underexplored underlying inflammation. Studies suggest a protective role for helminth infection in the prevention of T2D. The mechanisms may involve induction of modified type 2 and regulatory immune responses that suppress inflammation and promote insulin sensitivity. In this review, the roles of helminths in counteracting MetS, and prospects for harnessing these protective mechanisms for the development of novel anti-diabetes drugs are discussed.


Sujet(s)
Diabète de type 2 , Helminthes , Syndrome métabolique X , Animaux , Humains , Helminthes/immunologie , Helminthes/physiologie , Syndrome métabolique X/immunologie , Syndrome métabolique X/métabolisme , Syndrome métabolique X/parasitologie , Diabète de type 2/immunologie , Diabète de type 2/métabolisme , Helminthiase/immunologie , Helminthiase/parasitologie , Obésité/immunologie , Obésité/métabolisme , Interactions hôte-parasite/immunologie , Insulinorésistance
19.
Front Immunol ; 15: 1342431, 2024.
Article de Anglais | MEDLINE | ID: mdl-38655255

RÉSUMÉ

Chagas disease, caused by Trypanosoma cruzi, remains a serious public health problem worldwide. The parasite was subdivided into six distinct genetic groups, called "discrete typing units" (DTUs), from TcI to TcVI. Several studies have indicated that the heterogeneity of T. cruzi species directly affects the diversity of clinical manifestations of Chagas disease, control, diagnosis performance, and susceptibility to treatment. Thus, this review aims to describe how T. cruzi genetic diversity influences the biology of the parasite and/or clinical parameters in humans. Regarding the geographic dispersion of T. cruzi, evident differences were observed in the distribution of DTUs in distinct areas. For example, TcII is the main DTU detected in Brazilian patients from the central and southeastern regions, where there are also registers of TcVI as a secondary T. cruzi DTU. An important aspect observed in previous studies is that the genetic variability of T. cruzi can impact parasite infectivity, reproduction, and differentiation in the vectors. It has been proposed that T. cruzi DTU influences the host immune response and affects disease progression. Genetic aspects of the parasite play an important role in determining which host tissues will be infected, thus heavily influencing Chagas disease's pathogenesis. Several teams have investigated the correlation between T. cruzi DTU and the reactivation of Chagas disease. In agreement with these data, it is reasonable to suppose that the immunological condition of the patient, whether or not associated with the reactivation of the T. cruzi infection and the parasite strain, may have an important role in the pathogenesis of Chagas disease. In this context, understanding the genetics of T. cruzi and its biological and clinical implications will provide new knowledge that may contribute to additional strategies in the diagnosis and clinical outcome follow-up of patients with Chagas disease, in addition to the reactivation of immunocompromised patients infected with T. cruzi.


Sujet(s)
Maladie de Chagas , Variation génétique , Trypanosoma cruzi , Trypanosoma cruzi/génétique , Humains , Maladie de Chagas/immunologie , Maladie de Chagas/parasitologie , Animaux , Interactions hôte-parasite/génétique , Interactions hôte-parasite/immunologie
20.
Bull Math Biol ; 86(6): 62, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38662120

RÉSUMÉ

Hosts can evolve a variety of defences against parasitism, including resistance (which prevents or reduces the spread of infection) and tolerance (which protects against virulence). Some organisms have evolved different levels of tolerance at different life-stages, which is likely to be the result of coevolution with pathogens, and yet it is currently unclear how coevolution drives patterns of age-specific tolerance. Here, we use a model of tolerance-virulence coevolution to investigate how age structure influences coevolutionary dynamics. Specifically, we explore how coevolution unfolds when tolerance and virulence (disease-induced mortality) are age-specific compared to when these traits are uniform across the host lifespan. We find that coevolutionary cycling is relatively common when host tolerance is age-specific, but cycling does not occur when tolerance is the same across all ages. We also find that age-structured tolerance can lead to selection for higher virulence in shorter-lived than in longer-lived hosts, whereas non-age-structured tolerance always leads virulence to increase with host lifespan. Our findings therefore suggest that age structure can have substantial qualitative impacts on host-pathogen coevolution.


Sujet(s)
Évolution biologique , Interactions hôte-pathogène , Concepts mathématiques , Virulence , Animaux , Facteurs âges , Modèles biologiques , Interactions hôte-parasite/immunologie , Coévolution biologique , Humains , Longévité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE